Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Stroke ; 55(5): 1370-1380, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38572656

RESUMO

BACKGROUND: Mild chemical inhibition of mitochondrial respiration can confer resilience against a subsequent stroke or myocardial infarction, also known as preconditioning. However, the lack of chemicals that can safely inhibit mitochondrial respiration has impeded the clinical translation of the preconditioning concept. We previously showed that meclizine, an over-the-counter antivertigo drug, can toggle metabolism from mitochondrial respiration toward glycolysis and protect against ischemia-reperfusion injury in the brain, heart, and kidney. Here, we examine the mechanism of action of meclizine and report the efficacy and improved safety of the (S) enantiomer. METHODS: We determined the anoxic depolarization latency, tissue and neurological outcomes, and glucose uptake using micro-positron emission tomography after transient middle cerebral artery occlusion in mice pretreated (-17 and -3 hours) with either vehicle or meclizine. To exclude a direct effect on tissue excitability, we also examined spreading depression susceptibility. Furthermore, we accomplished the chiral synthesis of (R)- and (S)-meclizine and compared their effects on oxygen consumption and histamine H1 receptor binding along with their brain concentrations. RESULTS: Micro-positron emission tomography showed meclizine increases glucose uptake in the ischemic penumbra, providing the first in vivo evidence that the neuroprotective effect of meclizine indeed stems from its ability to toggle metabolism toward glycolysis. Consistent with reduced reliance on oxidative phosphorylation to sustain the metabolism, meclizine delayed anoxic depolarization onset after middle cerebral artery occlusion. Moreover, the (S) enantiomer showed reduced H1 receptor binding, a dose-limiting side effect for the racemate, but retained its effect on mitochondrial respiration. (S)-meclizine was at least as efficacious as the racemate in delaying anoxic depolarization onset and decreasing infarct volumes after middle cerebral artery occlusion. CONCLUSIONS: Our data identify (S)-meclizine as a promising new drug candidate with high translational potential as a chemical preconditioning agent for preemptive prophylaxis in patients with high imminent stroke or myocardial infarction risk.

2.
AJR Am J Roentgenol ; 220(5): 619-629, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36321986

RESUMO

Theranostics describes the coupling of a diagnostic biomarker and a therapeutic agent (i.e., a theranostic pair) that have a common target in tumor cells or their microenvironment. The term is increasingly associated with in vivo nuclear medicine oncologic applications that couple diagnostic imaging by means of gamma radiation with concomitant localized high-energy particulate radiation to a tissue expressing the common target. Several theranostic pairs have been translated into clinical practice in the United States and are poised to become a mainstay of cancer treatment. The purposes of this article are to review experience with theranostics for solid-organ malignancies and to address the practical integration into care pathways of ß-emitting therapies that include somatostatin analogue radioligands for neuroendocrine tumors, PSMA-directed therapy for prostate cancer, and 131I-MIBG therapy for tumors of neural crest origin. Toxicities related to theranostics administration and indications for cessation of therapy in patients who experience adverse events are also discussed. A multidisciplinary team-based approach for identifying patients most likely to respond to these agents, determining the optimal time for therapy delivery, and managing patient care throughout the therapeutic course is critical to the success of a radiotheranostic program.


Assuntos
Medicina de Precisão , Neoplasias da Próstata , Masculino , Humanos , Procedimentos Clínicos , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/radioterapia , Somatostatina , Assistência ao Paciente , Microambiente Tumoral
4.
Eur J Nucl Med Mol Imaging ; 48(5): 1522-1537, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33619599

RESUMO

BACKGROUND: MR is an important imaging modality for evaluating musculoskeletal malignancies owing to its high soft tissue contrast and its ability to acquire multiparametric information. PET provides quantitative molecular and physiologic information and is a critical tool in the diagnosis and staging of several malignancies. PET/MR, which can take advantage of its constituent modalities, is uniquely suited for evaluating skeletal metastases. We reviewed the current evidence of PET/MR in assessing for skeletal metastases and provided recommendations for its use. METHODS: We searched for the peer reviewed literature related to the usage of PET/MR in the settings of osseous metastases. In addition, expert opinions, practices, and protocols of major research institutions performing research on PET/MR of skeletal metastases were considered. RESULTS: Peer-reviewed published literature was included. Nuclear medicine and radiology experts, including those from 13 major PET/MR centers, shared the gained expertise on PET/MR use for evaluating skeletal metastases and contributed to a consensus expert opinion statement. [18F]-FDG and non [18F]-FDG PET/MR may provide key advantages over PET/CT in the evaluation for osseous metastases in several primary malignancies. CONCLUSION: PET/MR should be considered for staging of malignancies where there is a high likelihood of osseous metastatic disease based on the characteristics of the primary malignancy, hight clinical suspicious and in case, where the presence of osseous metastases will have an impact on patient management. Appropriate choice of tumor-specific radiopharmaceuticals, as well as stringent adherence to PET and MR protocols, should be employed.


Assuntos
Prova Pericial , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Fluordesoxiglucose F18 , Humanos , Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos
5.
Eur J Nucl Med Mol Imaging ; 47(1): 105-114, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31492992

RESUMO

PURPOSE: 18F-Fluciclovine is indicated for evaluation of suspected prostate cancer (PCa) biochemical recurrence. There are few studies investigating fluciclovine with PET/MR and none evaluated osseous metastases. Our aim was to assess the performance of 18F-fluciclovine PET/MR (fluciclovine-PET/MR) for detecting osseous metastases in patients with castration-resistant prostate cancer (CRPC). We also investigated possible correlations between SUVmax and ADCmean. METHODS: We evaluated 8 patients with CRPC metastatic to bones, some before and some after radium therapy, who underwent 13 fluciclovine-PET/MR studies. We analyzed the performance of radionuclide bone scan (RBS), MR alone, fluciclovine-PET alone, and fluciclovine-PET/MR in detecting osseous metastases. Lesion size, characteristics (early sclerotic, late sclerotic, mixed, lytic), SUVmax, and ADCmean were assessed. The reference standard was a combination of clinical information and correlation with both prior and follow-up imaging. RESULTS: Of 347 metastatic bony lesions in 13 studies, 238/347 (68%) were detected by fluciclovine-PET alone, 286/347 (82%) by RBS, 344/347 (99%) by MR alone, and 347/347 (100%) by fluciclovine-PET/MR. Fluciclovine-PET/MR and MR had the best performance (p < 0.001). There was no statistically significant difference between fluciclovine-PET/MR and MR alone (p = 0.25). Fluciclovine-PET had a lower detection rate especially with late sclerotic lesions (p < 0.001). There was a moderate inverse correlation between lesion SUVmax and ADCmean (r = - 0.49; p < 0.001). CONCLUSIONS: This study suggests that fluciclovine-PET/MR and MR have high sensitivity for detecting osseous metastases in CRPC. Fluciclovine-PET alone underperformed in detecting late sclerotic lesions. The inverse correlation between SUVmax and ADCmean suggests a possible relationship between tumor metabolism and cellularity.


Assuntos
Neoplasias Ósseas , Neoplasias de Próstata Resistentes à Castração , Neoplasias da Próstata , Neoplasias Ósseas/diagnóstico por imagem , Osso e Ossos , Humanos , Masculino , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias de Próstata Resistentes à Castração/diagnóstico por imagem
7.
Mol Imaging ; 13: 1-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24824854

RESUMO

Esophageal tumors provide unique challenges and opportunities for developing and testing surveillance imaging technology for different tumor microenvironment components, including assessment of immune cell modulation, with the ultimate goal of promoting early detection and response evaluation. In this context, accessibility through the lumen using a minimally invasive approach provides a means for repetitive evaluation longitudinally by combining fluorescent endoscopic imaging technology with novel fluorescent nanoparticles that are phagocytized by immune cells in the microenvironment. The agent we developed for imaging is synthesized from Feraheme (ferumoxytol), a Food and Drug Administration-approved monocrystaline dextran-coated iron oxide nanoparticle, which we conjugated to a near-infrared fluorochrome, CyAL5.5. We demonstrate a high level of uptake of the fluorescent nanoparticles by myeloid-derived suppressor cells (MDSCs) in the esophagus and spleen of L2Cre;p120ctnflox/flox mice. These mice develop esophageal dysplasia leading to squamous cell carcinoma; we have previously demonstrated that dysplastic and neoplastic esophageal lesions in these mice have an immune cell infiltration that is dominated by MDSCs. In the L2Cre;p120ctnflox/flox mice, evaluation of the spleen reveals that nearly 80% of CD45+ leukocytes that phagocytized the nanoparticle were CD11b+Gr1+ MDSCs. After dexamethasone treatment, we observed concordant decreased fluorescent signal from esophageal lesions during fluorescent endoscopy and decreased CyAL5.5-fluorescent-positive immune cell infiltration in esophageal dysplastic lesions by fluorescence-activated cell sorting analysis. Our observations suggest that this translatable technology may be used for the early detection of dysplastic changes and the serial assessment of immunomodulatory therapy and to visualize changes in MDSCs in the esophageal tumor microenvironment.


Assuntos
Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/cirurgia , Neoplasias Esofágicas/imunologia , Neoplasias Esofágicas/cirurgia , Óxido Ferroso-Férrico/química , Antígenos Comuns de Leucócito/imunologia , Animais , Antineoplásicos Hormonais/administração & dosagem , Carbocianinas/farmacocinética , Carcinoma de Células Escamosas/diagnóstico , Células Cultivadas , Dexametasona/administração & dosagem , Dimetil Sulfóxido/farmacologia , Endoscopia , Endoscopia Gastrointestinal , Neoplasias Esofágicas/diagnóstico , Óxido Ferroso-Férrico/farmacocinética , Corantes Fluorescentes/farmacocinética , Indóis/farmacocinética , Leucócitos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Nanopartículas/química , Baço/imunologia
8.
Gastroenterology ; 144(2): 294-297, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23085486

RESUMO

Imaging strategies that detect early stage esophageal squamous cell carcinoma (ESCC) could improve clinical outcomes, when combined with endoscopic approaches. Periostin is an integrin-binding protein that is important in the tumor microenvironment. We created a fluorescent-labeled antibody that recognizes periostin and binds specifically to ESCC xenograft tumors in mice. In L2-cre;p120ctnLoxP/LoxP mice, which develop squamous cell cancers that resemble human ESCC, we visualized the probe in preneoplastic and neoplastic esophageal lesions using near-infrared fluorescent imaging with upper-gastrointestinal endoscopy. Periostin might be a biomarker of the esophageal tumor microenvironment that can be used to detect preneoplastic lesions.


Assuntos
Carcinoma de Células Escamosas/diagnóstico , Diagnóstico Precoce , Neoplasias Esofágicas/diagnóstico , Esofagoscopia/métodos , Neoplasias Experimentais , Imagem Óptica/métodos , Animais , Diagnóstico Diferencial , Humanos , Camundongos
9.
Radiology ; 271(3): 770-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24520946

RESUMO

PURPOSE: To investigate indocyanine green (ICG) as a molecular beacon for malignant lesions within the liver and evaluate the ability of a developed handheld imaging system to allow measurement of ICG fluorescence within focal hepatic lesions with high target-to-background ratios in a mouse model. MATERIALS AND METHODS: All animal experiments were approved by the institutional animal care committee. A handheld optical molecular imaging device was constructed to pass through the introducer needle of a standard percutaneous biopsy kit. An ex vivo phantom system was constructed to quantify tissue attenuation properties of ICG in liver parenchyma. Subsequently, intrahepatic colorectal cancer metastases were generated in nude mice, and epifluorescence imaging of ICG, as well as histologic analysis of the explanted livers, was performed at 3 weeks after implantation (n = 6). Epifluorescence imaging with the handheld imaging device was then performed on intrahepatic colorectal metastases after the administration of ICG (n = 15) at 3, 6, and 24 hours after injection. Target-to-background ratios were calculated for each time point. Subsequently, a core biopsy of intrahepatic colorectal metastases was performed by using a standard clinical 18-gauge biopsy needle. RESULTS: There was avid localization of ICG to the focal lesions at all time points. Similarly, fluorescence within the tumors was greater than that within normal liver, as detected with the handheld imaging system (mean target-to-background ratio ± standard deviation, 3.9 ± 0.2 at 24 hours). A core biopsy of tumor and normal adjacent liver by using a standard biopsy needle demonstrated a sharp margin of fluorescence intensity at the tumor-liver interface. CONCLUSION: The custom-designed molecular imaging device, in combination with ICG, readily allowed differentiation between normal and malignant tissue in a murine model of intrahepatic colorectal metastasis.


Assuntos
Biópsia/métodos , Neoplasias Hepáticas Experimentais/patologia , Imagem Molecular/métodos , Animais , Corantes , Verde de Indocianina , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Imagem Molecular/instrumentação , Imagens de Fantasmas
10.
Stem Cells ; 31(8): 1706-14, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23389839

RESUMO

Therapeutically engineered stem cells (SC) are emerging as an effective tumor-targeted approach for different cancer types. However, the assessment of the long-term fate of therapeutic SC post-tumor treatment is critical if such promising therapies are to be translated into clinical practice. In this study, we have developed an efficient SC-based therapeutic strategy that simultaneously allows killing of tumor cells and assessment and eradication of SC after treatment of highly malignant glioblastoma multiforme (GBM). Mesenchymal stem cells (MSC) engineered to co-express the prodrug converting enzyme, herpes simplex virus thymidine kinase (HSV-TK) and a potent and secretable variant of tumor necrosis factor apoptosis-inducing ligand (S-TRAIL) induced caspase-mediated GBM cell death and showed selective MSC sensitization to the prodrug ganciclovir (GCV). A significant decrease in tumor growth and a subsequent increase in survival were observed when mice bearing highly aggressive GBM were treated with MSC coexpressing S-TRAIL and HSV-TK. Furthermore, the systemic administration of GCV post-tumor treatment selectively eliminated therapeutic MSC expressing HSV-TK in vitro and in vivo, which was monitored in real time by positron emission-computed tomography imaging using 18F-FHBG, a substrate for HSV-TK. These findings demonstrate the development and validation of a novel therapeutic strategy that has implications in translating SC-based therapies in cancer patients.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Animais , Apoptose/fisiologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Ganciclovir/administração & dosagem , Ganciclovir/farmacocinética , Engenharia Genética/métodos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/cirurgia , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Simplexvirus/enzimologia , Simplexvirus/genética , Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Ligante Indutor de Apoptose Relacionado a TNF/genética , Timidina Quinase/biossíntese , Timidina Quinase/genética
11.
J Nucl Med ; 65(1): 4-12, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37945384

RESUMO

Nuclear medicine imaging modalities such as PET and SPECT are confounded by high noise levels and low spatial resolution, necessitating postreconstruction image enhancement to improve their quality and quantitative accuracy. Artificial intelligence (AI) models such as convolutional neural networks, U-Nets, and generative adversarial networks have shown promising outcomes in enhancing PET and SPECT images. This review article presents a comprehensive survey of state-of-the-art AI methods for PET and SPECT image enhancement and seeks to identify emerging trends in this field. We focus on recent breakthroughs in AI-based PET and SPECT image denoising and deblurring. Supervised deep-learning models have shown great potential in reducing radiotracer dose and scan times without sacrificing image quality and diagnostic accuracy. However, the clinical utility of these methods is often limited by their need for paired clean and corrupt datasets for training. This has motivated research into unsupervised alternatives that can overcome this limitation by relying on only corrupt inputs or unpaired datasets to train models. This review highlights recently published supervised and unsupervised efforts toward AI-based PET and SPECT image enhancement. We discuss cross-scanner and cross-protocol training efforts, which can greatly enhance the clinical translatability of AI-based image enhancement tools. We also aim to address the looming question of whether the improvements in image quality generated by AI models lead to actual clinical benefit. To this end, we discuss works that have focused on task-specific objective clinical evaluation of AI models for image enhancement or incorporated clinical metrics into their loss functions to guide the image generation process. Finally, we discuss emerging research directions, which include the exploration of novel training paradigms, curation of larger task-specific datasets, and objective clinical evaluation that will enable the realization of the full translation potential of these models in the future.


Assuntos
Inteligência Artificial , Processamento de Imagem Assistida por Computador , Processamento de Imagem Assistida por Computador/métodos , Razão Sinal-Ruído , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia por Emissão de Pósitrons/métodos
12.
J Nucl Med ; 65(7): 1137-1143, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38754959

RESUMO

Developing a noninvasive imaging method to detect immune system activation with a high temporal resolution is key to improving inflammatory bowel disease (IBD) management. In this study, granzyme B (GZMB), typically released from cytotoxic T and natural killer cells, was targeted using PET with 68Ga-NOTA-GZP (where GZP is ß-Ala-Gly-Gly-Ile-Glu-Phe-Asp-CHO) to detect early intestinal inflammation in murine models of colitis. Methods: Bioinformatic analysis was used to assess the potential of GZMB as a biomarker for detecting IBD and predicting response to treatment. Human active and quiescent Crohn disease and ulcerative colitis tissues were stained for GZMB. We used IL-10-/- mice treated with dextran sulfate sodium (DSS) as an IBD model, wild-type C57BL/6J mice as a control, and anti-tumor necrosis factor as therapy. We used a murine GZMB-binding peptide conjugated to a NOTA chelator (NOTA-GZP) labeled with 68Ga as the PET tracer. PET imaging was conducted at 1, 3, and 4 wk after colitis induction to evaluate temporal changes. Results: Bioinformatic analysis showed that GZMB gene expression is significantly upregulated in human ulcerative colitis and Crohn disease compared with the noninflamed bowel by 2.98-fold and 1.92-fold, respectively; its expression is lower by 2.16-fold in treatment responders than in nonresponders. Immunofluorescence staining of human tissues demonstrated a significantly higher GZMB in patients with active than with quiescent IBD (P = 0.032).68Ga-NOTA-GZP PET imaging showed significantly increased bowel uptake in IL-10-/- mice with DSS-induced colitis compared with vehicle-treated IL-10-/- mice (SUVmean, 0.75 vs. 0.24; P < 0.001) and both vehicle- and DSS-treated wild-type mice (SUVmean, 0.26 and 0.37; P < 0.001). In the IL-10-/- DSS-induced colitis model, the bowel PET probe uptake decreased in response to treatment with tumor necrosis factor-α (SUVmean, 0.32; P < 0.001). There was a 4-fold increase in colonic uptake of 68Ga-NOTA-GZP in the colitis model compared with the control 1 wk after colitis induction. The uptake gradually decreased to approximately 2-fold by 4 wk after IBD induction; however, the inflamed bowel uptake remained significantly higher than control at all time points (week 4 SUVmean, 0.23 vs. 0.08; P = 0.001). Conclusion: GZMB is a promising biomarker to detect active IBD and predict response to treatment. This study provides compelling evidence to translate GZMB PET for imaging IBD activity in clinical settings.


Assuntos
Granzimas , Doenças Inflamatórias Intestinais , Tomografia por Emissão de Pósitrons , Animais , Camundongos , Doenças Inflamatórias Intestinais/diagnóstico por imagem , Humanos , Granzimas/metabolismo , Camundongos Endogâmicos C57BL
13.
Urol Clin North Am ; 50(1): 115-131, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36424076

RESUMO

Urologic malignancies constitute a large portion of annually diagnosed cancers. Timely diagnosis, accurate staging, and assessment of tumor heterogeneity are essential to devising the best treatment strategy for individual patients. The high sensitivity of molecular imaging allows for early and sensitive detection of lesions that were not readily detectable using conventional imaging techniques. Moreover, molecular imaging enables the interrogation of molecular processes used in targeted cancer therapies and predicts cancer response to treatment. Here we review the current advancements in molecular imaging of urologic cancers, including prostatic, vesical, renal testicular, and ureteral cancers.


Assuntos
Neoplasias Urológicas , Humanos , Neoplasias Urológicas/diagnóstico por imagem , Neoplasias Urológicas/patologia , Biomarcadores , Imagem Molecular
14.
Life Sci ; 329: 121970, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37481033

RESUMO

Cancer cells are surrounded by a complex and highly dynamic tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs), a critical component of TME, contribute to cancer cell proliferation as well as metastatic spread. CAFs express a variety of biomarkers, which can be targeted for detection and therapy. Most importantly, CAFs express high levels of fibroblast activation protein (FAP) which contributes to progression of cancer, invasion, metastasis, migration, immunosuppression, and drug resistance. As a consequence, FAP is an attractive theranostic target. In this review, we discuss the latest advancement in targeting FAP in oncology using theranostic biomarkers and imaging modalities such as single-photon emission computed tomography (SPECT), positron emission tomography (PET), computed tomography (CT), fluorescence imaging, and magnetic resonance imaging (MRI).


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Humanos , Serina Endopeptidases/metabolismo , Medicina de Precisão , Proteínas de Membrana/metabolismo , Neoplasias/terapia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos/metabolismo , Microambiente Tumoral
15.
Optics (Basel) ; 4(2): 340-350, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38075027

RESUMO

Image-guided liver biopsies can improve their success rate when combined with the optical detection of Indocyanine Green (ICG) fluorescence accumulated in tumors. Previous works used a camera coupled to a thin borescope to capture and quantify images from fluorescence emission during procedures; however, light-scattering prevented the formation of sharp images, and the time response for weakly fluorescent tumors was very low. Instead, replacing the camera with a photodiode detector shows an improved temporal resolution in a more compact and lighter device. This work presents the new design in a comparative study between both detection technologies, including an assessment of the temporal response and sensitivity to the presence of background fluorescence.

16.
J Nucl Med ; 64(7): 1056-1061, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37024303

RESUMO

Neuroendocrine tumors (NETs) are often diagnosed in advanced stages. Despite the advances in treatment approaches, including somatostatin analogs and peptide receptor radionuclide therapy (PRRT), these patients have no curative treatment option. Moreover, immunotherapy often yields modest results in NETs. We investigated whether combining PRRT using [177Lu]DOTATATE and immune checkpoint inhibition therapy improves treatment response in NETs. Methods: A gastroenteropancreatic NET model was generated by subcutaneous implantation of human QGP-1 cells in immune-reconstituted NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ mice engrafted with human peripheral blood mononuclear cells (n = 96). Mice were randomly assigned to receive pembrolizumab (anti-PD1), [177Lu]DOTATATE (PRRT), simultaneous anti-PD1 and PRRT (S-PRRT), anti-PD1 on day 0 followed by PRRT on day 3 (delayed PRRT [D-PRRT]), PRRT on day 0 followed by anti-PD1 (early PRRT [E-PRRT]), or vehicle as control (n = 12/group). Human granzyme-B-specific [68Ga]NOTA-hGZP PET/MRI was performed before and 6 d after treatment initiation, as an indicator of T-cell activation. Response to treatment was based on tumor growth over 21 d and on histologic analyses of extracted tissues on flow cytometry for T cells, hematoxylin and eosin staining, and immunohistochemical staining. Results: [68Ga]NOTA-hGZP PET/MRI showed significantly increased uptake in tumors treated with E-PRRT, S-PRRT, and anti-PD1 on day 6 compared with baseline (SUVmax: 3.36 ± 0.42 vs. 0.73 ± 0.23; 2.36 ± 0.45 vs. 0.76 ± 0.30; 2.20 ± 0.20 vs. 0.72 ± 0.28, respectively; P < 0.001), whereas no significant change was seen in PET parameters in the D-PRRT, PRRT, or vehicle groups (P > 0.05). Ex vivo analyses confirmed the PET results showing the highest granzyme-B levels and T cells (specifically CD8-positive effector T cells) in the E-PRRT group, followed by the S-PRRT and anti-PD1 groups. Tumor growth follow-up showed the most significant tumor size reduction in the E-PRRT group (baseline to day 21, 205.00 ± 30.70 mm3 vs. 78.00 ± 11.75 mm3; P = 0.0074). Tumors showed less growth reduction in the PRRT, D-PRRT, and S-PRRT groups than in the E-PRRT group (P < 0.0001). The vehicle- and anti-PD-1-treated tumors showed continued growth. Conclusion: Combination of PRRT and anti-PD1 shows the most robust inflammatory response to NETs and a better overall outcome than immune checkpoint inhibition or PRRT alone. The most effective regimen is PRRT preceding anti-PD1 administration by several days.


Assuntos
Tumores Neuroendócrinos , Compostos Organometálicos , Humanos , Animais , Camundongos , Granzimas , Inibidores de Checkpoint Imunológico , Tumores Neuroendócrinos/diagnóstico por imagem , Tumores Neuroendócrinos/radioterapia , Tumores Neuroendócrinos/patologia , Radioisótopos de Gálio , Leucócitos Mononucleares/patologia , Camundongos Endogâmicos NOD , Receptores de Peptídeos , Octreotida
17.
IEEE Trans Med Imaging ; PP2023 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-37995174

RESUMO

Position emission tomography (PET) is widely used in clinics and research due to its quantitative merits and high sensitivity, but suffers from low signal-to-noise ratio (SNR). Recently convolutional neural networks (CNNs) have been widely used to improve PET image quality. Though successful and efficient in local feature extraction, CNN cannot capture long-range dependencies well due to its limited receptive field. Global multi-head self-attention (MSA) is a popular approach to capture long-range information. However, the calculation of global MSA for 3D images has high computational costs. In this work, we proposed an efficient spatial and channel-wise encoder-decoder transformer, Spach Transformer, that can leverage spatial and channel information based on local and global MSAs. Experiments based on datasets of different PET tracers, i.e., 18F-FDG, 18F-ACBC, 18F-DCFPyL, and 68Ga-DOTATATE, were conducted to evaluate the proposed framework. Quantitative results show that the proposed Spach Transformer framework outperforms state-of-the-art deep learning architectures.

18.
Clin Imaging ; 99: 10-18, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37043868

RESUMO

COVID-19 is a multisystemic disease, and hence its potential manifestations on nuclear medicine imaging can extend beyond the lung. Therefore, it is important for the nuclear medicine physician to recognize these manifestations in the clinic. While FDG-PET/CT is not indicated routinely in COVID-19 evaluation, its unique capability to provide a functional and anatomical assessment of the entire body means that it can be a powerful tool to monitor acute, subacute, and long-term effects of COVID-19. Single-photon scintigraphy is routinely used to assess conditions such as pulmonary embolism, cardiac ischemia, and thyroiditis, and COVID-19 may present in these studies. The most common nuclear imaging finding of COVID-19 vaccination to date is hypermetabolic axillary lymphadenopathy. This may pose important diagnostic and management dilemmas in oncologic patients, particularly those with malignancies where the axilla constitutes a lymphatic drainage area. This article aims to summarize the relevant literature published since the beginning of the pandemic on the intersection between COVID-19 and nuclear medicine.


Assuntos
COVID-19 , Medicina Nuclear , Humanos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Vacinas contra COVID-19 , Fluordesoxiglucose F18 , Cintilografia , Tomografia por Emissão de Pósitrons , Dedos do Pé
19.
Pharmaceutics ; 15(12)2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38140100

RESUMO

DNA is an organic molecule that is highly vulnerable to chemical alterations and breaks caused by both internal and external factors. Cells possess complex and advanced mechanisms, including DNA repair, damage tolerance, cell cycle checkpoints, and cell death pathways, which together minimize the potentially harmful effects of DNA damage. However, in cancer cells, the normal DNA damage tolerance and response processes are disrupted or deregulated. This results in increased mutagenesis and genomic instability within the cancer cells, a known driver of cancer progression and therapeutic resistance. On the other hand, the inherent instability of the genome in rapidly dividing cancer cells can be exploited as a tool to kill by imposing DNA damage with radiopharmaceuticals. As the field of targeted radiopharmaceutical therapy (RPT) is rapidly growing in oncology, it is crucial to have a deep understanding of the impact of systemic radiation delivery by radiopharmaceuticals on the DNA of tumors and healthy tissues. The distribution and activation of DNA damage and repair pathways caused by RPT can be different based on the characteristics of the radioisotope and molecular target. Here we provide a comprehensive discussion of the biological effects of RPTs, with the main focus on the role of varying radioisotopes in inducing direct and indirect DNA damage and activating DNA repair pathways.

20.
Mol Imaging Biol ; 25(2): 353-362, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35962301

RESUMO

PURPOSE: New generation of receptor tyrosine kinase inhibitors (RTKIs) have shown to improve survival in many solid tumors. However, an imaging biomarker is needed for patient selection and prediction of treatment response. This study evaluates the use of quantitative changes of HER3 on 68 Ga-NOTA-HER3P1 PET/MRI for prediction of early response to pan-RTKIs in gastric cancer (GCa). PROCEDURES: GCa cell lines were evaluated for expression of RTKs, and downstream signaling pathways (AKT and MAPK). Cell viability was assessed following 24-72 h of treatment with 0.01-1 µmol/L of afatinib, a pan-RTKI. HER3-expressing afatinib-sensitive (NCI-N87) and resistant cells (SNU16) were selected for evaluation of changes in RTKs expression and downstream pathways, with 24-72 h of 0.1 µmol/L afatinib treatment. 68 Ga-NOTA-HER3P1 PET/MRI was performed in subcutaneous NCI-N87 and SNU16 xenografts (nu:nu, n = 12/group) at baseline and 4 days after afatinib treatment (10 mg/kg, PO, daily). Temporal changes in PET measures were correlated to HER3 expression in tumors, tumor growth rate, and treatment response. RESULTS: With afatinib therapy, NCI-N87 cells showed increased total HER3 expression, and reduction of other RTKs and downstream nodes within 72 h, while SNU16 cells showed no significant change in total HER3 and downstream nodes. 68 Ga-HER3P1 PET/MRI showed increased uptake in NCI-N87 and no significant change in SNU16 tumors (day 4 vs. baseline SUVmean: 3.8 ± 0.7 vs. 1.6 ± 0.6, p < 0.05 in NCI-N87, and 1.5 ± 0.7 vs. 1.7 ± 0.7, p > 0.05 in SNU16). These findings were in concordance with HER3 expression in histopathological analyses and tumor growth over 3 weeks of treatment (mean tumor volume in treated vs. control: 11 ± 17 mm3 vs. 293 ± 79 mm3, p < 0.001 in NCI-N87, and 238 ± 91 mm3 vs. 282 ± 35 mm3, p > 0.05 in SNU16). CONCLUSIONS: Quantitative changes in HER3 PET could be used to predict response to pan-RTKI within few days after initiation of treatment and can help with personalizing GCa management.


Assuntos
Neoplasias Gástricas , Humanos , Afatinib/farmacologia , Neoplasias Gástricas/patologia , Linhagem Celular Tumoral , Tomografia por Emissão de Pósitrons/métodos , Receptor ErbB-3
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa